throbber
Core Entrustables in Clinical Pharmacology: Pearls for Clinical Practice
`
`Pharmacokinetics and Pharmacodynamics
`for Medical Students: A Proposed
`Course Outline
`
`The Journal of Clinical Pharmacology
`2016, 56(10) 1180–1195
`C(cid:2) 2016, The American College of
`Clinical Pharmacology
`DOI: 10.1002/jcph.732
`
`David J. Greenblatt, MD,1 and Paul N. Abourjaily, PharmD2
`
`Keywords
`pharmacokinetics, pharmacodynamics, drug interactions, medical
`
`The discipline of pharmacokinetics (PK) applies math-
`ematical models to describe and predict the time
`course of drug concentrations and drug amounts in
`body fluids.1–3 Pharmacodynamics (PD) applies sim-
`ilar models to understand the time course of drug
`actions on the body. Clinicians are most concerned with
`pharmacodynamics—they want to know how drug
`dosage, route of administration, and frequency of ad-
`ministration can be chosen to maximize the probability
`of therapeutic success while minimizing the likelihood
`of unwanted drug effects. However the path to pharma-
`codynamics comes via pharmacokinetics. Because drug
`effects are related to drug concentrations, understand-
`ing and predicting the time course of concentrations
`can be used to help optimize therapy.
`The link of drug dosage to drug effect involves
`a sequence of events (Figure 1). Even when a drug
`is administered directly into the vascular system, the
`drug diffuses to both its pharmacologic target receptor
`and to other peripheral distribution sites where it does
`not have the desired activity but may exert toxic ef-
`fects. Simultaneously, the drug undergoes clearance by
`metabolism and excretion. After oral administration,
`the situation is more complex, since the drug must
`undergo dissolution and absorption, then survive first-
`pass metabolism in the liver, before reaching the sys-
`temic circulation. Pharmacokinetics provides a ratio-
`nal mathematical framework for understanding these
`concurrent processes, and facilitates achieving optimal
`clinical pharmacodynamic effects more efficiently than
`trial and error alone.
`The following 3 clinical vignettes illustrate how
`familiarity with principles of pharmacokinetics and
`pharmacodynamics can facilitate optimal understand-
`ing and prediction of drug effects in human subjects
`and patients.
`
`Clinical Vignettes
`Case 1
`A 30-year-old man has been extensively evaluated for
`recurrent supraventricular tachycardia (SVT), which is
`associated with palpitations and dizziness. No iden-
`tifiable cardiac disease is evident, and other medical
`diseases have been excluded.
`The treating physician elects to start therapy with
`digitoxin, 0.1 mg daily. One week later the patient is seen
`again, and states that episodes of SVT are reduced in
`number. The plasma digitoxin level is 8 ng/mL (usual
`therapeutic range, 10–20 ng/mL). The dose is increased
`to 0.2 mg/day. At a follow-up visit 7 days later, the
`patient claims that symptoms attributable to SVT have
`disappeared completely. The plasma digitoxin level is
`17.4 ng/mL. The patient continues on 0.2 mg/day of
`digitoxin.
`One month later the patient sees the physician on
`an urgent basis. He has diminished appetite and waves
`
`1Program in Pharmacology and Experimental Therapeutics, Sackler
`School of Graduate Biomedical Sciences, Tufts University School of
`Medicine, Boston, MA, USA
`2Departments of Pharmacy and Medicine, Tufts Medical Center, Boston,
`MA, USA
`
`Submitted for publication 26 February 2016; accepted 3 March 2016.
`
`Corresponding Author:
`David J. Greenblatt, MD, Tufts University School of Medicine, 136
`Harrison Avenue, Boston, MA 02111
`Email: DJ.Greenblatt@Tufts.edu
`
`This proposal was prepared under the guidance of the Special Com-
`mittee for Medical School Curriculum of the American College of
`Clinical Pharmacology. The proposal
`is intended as a resource for
`the Association of American Medical Colleges as it revises its Core
`Entrustable Professional Activities for Entering Residency, Curriculum
`Developer’s Guide.
`
`1
`
`TEVA1058
`
`

`

`Greenblatt and Abourjaily
`
`1181
`
`Figure 1. Sequence of events between intravenous or oral adminis-
`tration of a drug and the drug’s interaction with the target receptor
`mediating pharmacologic action.This type of schematic diagram has been
`attributed to Dr. Leslie Z. Benet. The segment above the dashed line is
`the pharmacokinetic component—“what the body does to the drug.”
`Below the dashed line is the pharmacodynamic component—“what the
`drug does to the body” (from reference 2, with permission).
`
`of nausea. The electrocardiogram shows T-wave abnor-
`malities, and the plasma digitoxin level is 31.3 ng/mL.
`
`Discussion—Case 1
`Digitoxin is seldom used in contemporary therapeutics,
`but case 1 nonetheless illustrates 2 important principles:
`dose proportionality, and attainment of steady-state.
`The rate of attainment of the steady-state condition
`after initiation of multiple-dose treatment is dependent
`on the half-life of the particular drug. In the case of
`digitoxin, the half-life is about 7 days, implying that 3–4
`weeks of continuous treatment (without a loading dose)
`is necessary for steady-state to be reached.4 In the exam-
`ple given, the increase in dosage from 0.1 to 0.2 mg/day
`is anticipated to proportionally increase the steady-
`state concentration (Figure 2). At the daily dosage of
`0.1 mg (without a loading dose) and a half-life of 7
`days, the plasma digitoxin concentration of 8 ng/mL
`after 7 days of treatment represents 50% of the eventual
`steady-state concentration of 16 ng/mL. If the physi-
`cian had stayed with the 0.1 mg/day dosage, that steady-
`state concentration—attained after several weeks of
`treatment (4 to 5 times the half-life)—would have been
`in the therapeutic range. However the dosage was in-
`creased to 0.2 mg/day, yielding a corresponding steady-
`state concentration of 32 ng/mL, exceeding the thera-
`peutic range and producing adverse effects (Figure 2).
`
`Case 2
`A 56-year-old man has a history of grand mal seizures,
`which have been completely suppressed for the last 12
`years by phenytoin, 300 mg daily. His plasma phenytoin
`level consistently falls in the range of 12–16 μg/mL. The
`patient is able to lead a normal life, and is an excellent
`tennis player. During a particularly competitive tennis
`match, the patient injures his shoulder. That night he
`experiences severe pain, tenderness, and limitation of
`
`Figure 2. Hypothetical mean plasma concentrations of digitoxin, cor-
`responding to case 1 in the text. Digitoxin is initially given at a dosage
`of 0.1 mg per day for 1 week, after which the plasma concentration
`is 8 ng/mL (point a). The treating physician wants to increase the
`plasma concentration to a value within the usual therapeutic range
`(10–20 ng/mL). If the physician stayed with the 0.1-mg-per-day dosage,
`the eventual steady-state concentration would have been 16 ng/mL
`(dashed line)—within the desired therapeutic range. Instead, the dosage
`is increased to 0.2 mg per day on day 7. The next measured plasma
`concentration is 17.4 ng/mL 1 week later (point b), but 1 month later
`it has reached 31.3 ng/mL (point c), close to the eventual steady-state
`value of 32 ng/mL. This is well above the therapeutic range and may be
`associated with toxicity.
`
`motion. He contacts his physician. An x-ray is negative.
`The physician prescribes rest, topical heat, and aspirin,
`650 mg 4 times daily.
`At a return visit to the physician 2 days later, the
`patient is greatly improved. The physician uses that
`opportunity to do a routine check of
`the plasma
`phenytoin level, which is reported as 5 μg/mL. There
`is no evidence of recurrent seizure activity, and the
`patient insists that he is continuing to take phenytoin
`as directed (300 mg/day). The physician increases the
`dose to 500 mg/day.
`One week later the patient returns complaining of
`difficulty with balance and with fixing his eyes on
`objects. The plasma phenytoin level is 14 μg/mL.
`
`Discussion—Case 2
`In case 2, plasma protein binding of phenytoin is
`reduced by coadministration of aspirin because of
`displacement of phenytoin from plasma-binding sites
`by salicylate.5–7 This is evident as an increase in the free
`fraction (Table 1). However, the clearance of unbound
`(free) drug, and the steady-state concentration of
`unbound drug, are unchanged.8,9 As such, no change
`in clinical effect would be anticipated, and the correct
`clinical course would have been to leave the daily dosage
`at 300 mg/day. Because salicylate reduces plasma
`protein binding of phenytoin (higher free fraction in
`plasma), this has the effect of reducing total (free +
`
`2
`
`

`

`1182
`
`The Journal of Clinical Pharmacology / Vol 56 No 10 2016
`
`Table 1. Total and Free (Unbound) Plasma Phenytoin Concentrations
`in Case 2
`
`Phenytoin Daily
`Dosage, and
`Cotreatment
`
`300 mg/day (no
`cotreatment)
`300 mg/day +
`aspirin
`500 mg/day +
`aspirin
`
`Total Phenytoin
`(μg/mL)
`
`Fraction Free
`
`12–16
`
`5
`
`14
`
`0.1
`
`0.3
`
`0.3
`
`Free Phenytoin
`(μg/mL)
`
`1.2–1.6
`
`1.5
`
`4.2
`
`bound) concentrations of phenytoin as well as the in-
`terpretation of these measured total concentrations.5–7
`Increasing the daily dosage of phenytoin to 500
`mg/day increases the free (unbound) concentration to
`4.2 μg/mL, which is associated with adverse effects
`despite the total concentration of 14 μg/mL.
`A second important point is the nonlinear kinetic
`profile of phenytoin.10 At daily doses in a range
`exceeding 300 mg/day, steady-state plasma concentra-
`tions increase disproportionately with an increase in
`dosage. The free phenytoin concentration is 1.5 μg/mL
`at 300 mg/day, but increases to 4.2 μg/mL with an
`increase in dosage to 500 mg/day. This property of
`phenytoin makes it difficult to titrate dosage at this
`higher dosage range.
`
`Case 3
`A fourth-year dental student is doing a clerkship in a
`dental surgeon’s practice. A healthy 34-year-old woman
`is scheduled to undergo procedures estimated to last
`approximately 2 hours. Following instillation of local
`anesthesia and prior to the start of the procedure,
`the surgeon notices that the patient still is extremely
`agitated and fearful. The surgeon administers 0.5 mg/kg
`of propofol intravenously over a 2-minute period. The
`patient becomes calm, relaxed, and falls into a light
`sleep from which she is easily roused. The surgical
`procedure is initiated and proceeds without incident for
`about 45 minutes. At this time, the patient becomes
`alert, and again is fearful and agitated. The surgeon ad-
`ministers another 0.5 mg/kg of propofol intravenously,
`the patient again becomes calm, and the surgical proce-
`dure proceeds to completion without incident.
`The student is confused. He/she asks the surgeon,
`“Why did the patient wake up after only 45 minutes?
`The half-life of propofol usually is at least 8 hours.”
`
`Discussion—Case 3
`Case 3 is an example of how the pharmacodynamic
`effects of lipophilic psychotropic drugs after single
`intravenous doses are dependent more on the rapid
`process of peripheral distribution than on clearance
`
`Figure 3. Hypothetical plasma concentrations of propofol correspond-
`ing to case 3 in the text. A 0.5 mg/kg intravenous dose of propofol is
`initially given at time zero. The plasma propofol concentration declines
`rapidly, falling below the hypothetical minimum effective concentration
`(MEC) of 200 ng/mL at 0.75 hours, at which time the patient emerges
`from the sedated condition. The rate of drug disappearance in the
`postdistributive phase would be slower (dashed line), but an additional
`0.5 mg/kg dose of propofol is required at the 0.75-hour time to maintain
`plasma concentrations above the MEC and maintain the patient in a
`sedated condition for the remainder of the procedure.
`
`or elimination. The pharmacokinetics of propofol are
`described by a 2- or 3-compartment model, in which
`the initial “distribution” phase represents rapid distri-
`bution from systemic circulation to peripheral tissues,
`followed by the terminal “elimination” phase which
`mainly reflects clearance.11–14 Although the propofol
`has not been eliminated from the body, its distribution
`from systemic circulation into peripheral tissue results
`in lower concentrations in plasma and brain, which is
`highly vascular and rapidly equilibrates with systemic
`circulation (Figure 3). As such, the patient becomes
`more alert, and a second dose is required.
`
`Components of Medical
`Education in Pharmacokinetics
`and Pharmacodynamics
`An outline of key elements of content for the teaching
`of clinical pharmacokinetics and pharmacodynamics
`to first- or second-year medical students, along with
`pertinent literature references,15–54 are presented in
`Table 2. The same or similar content can be applied to
`the teaching of graduate students at both the PhD and
`masters levels, or to postdoctoral education programs
`for house staff or practicing physicians. The material
`can be reasonably presented in 3 or 4 total lecture
`contact hours. The didactic presentations should be
`reinforced through problem sets and review sessions
`
`3
`
`

`

`Greenblatt and Abourjaily
`
`1183
`
`aimed at supporting conceptual understanding, as well
`as ensuring proficiency with pharmacokinetic calcula-
`tions and construction of graphics.
`Individual instructors can adapt the outline and
`accompanying graphics as needed, to construct specific
`lecture content consistent with their own style and
`institutional needs. An ongoing point of discussion is
`the extent to which formulas, equations, and mathemat-
`ics are needed for the teaching of pharmacokinetics.
`Student backgrounds in mathematics and their comfort
`with quantitative content vary widely. Some dislike
`and resist the mathematical content, whereas others
`welcome it. “Equation-free” pharmacokinetics is not
`realistic, but the density of equations can be managed
`such that the mathematical framework enhances con-
`
`ceptual understanding. The outline in Table 2 has that
`objective. The need for memorization of formulas and
`equations is minimal.
`Table 3 lists biomedical journals that have a focus
`on clinical pharmacokinetics and pharmacodynamics.
`Students are encouraged to consult original research
`sources when seeking information on pharmacoki-
`netic/pharmacodynamic properties or drug interactions
`involving specific drugs or drug classes. Review articles
`and secondary sources do have a role in the educational
`process, in that large amounts of data are collated and
`summarized. However, secondary sources are inevitably
`“filtered” and interpreted by their authors. Students
`need to consider the benefits and drawbacks of available
`information sources.
`
`4
`
`

`

`1184
`
`The Journal of Clinical Pharmacology / Vol 56 No 10 2016
`
`Table 2. Course Outline: Pharmacokinetics and Pharmacodynamics for a Medical School Curriculum
`
`Section 1. Definition and scope
`A. Pharmacokinetics: concentration versus time
`B. Pharmacodynamics: effect versus time
`C. Kinetic-dynamic modeling: effect versus concentration
`Section 2. Value of pharmacokinetic principles in medical science15–19
`A. Choice of loading and maintenance dose
`B. Choice of frequency and route of administration
`C. Predicting the rate and extent of drug accumulation
`D. Predicting the effect of dose changes
`E. Identifying and anticipating drug interactions
`F. Identifying patient and disease factors that could alter clinical response
`G. Interpreting drug concentrations in serum or plasma19
`
`Section 3. Fundamental assumptions
`A. Proportionality cascade (Figure 4):
`–Intravascular free drug
`–Extracellular water
`–Receptor occupancy
`–Quantitative pharmacodynamic effect
`B. Concentration ranges
`–Subtherapeutic
`–Therapeutic
`–Potentially toxic
`
`Section 4. Body compartments and volumes of distribution15–17,20–22
`A. Definition of a compartment
`B. Calculation of volume of distribution derived from definition of concentration:
`Concentration = Amount
`Volume
`Volume of distribution Vd =
`
`Amount of drug in body
`Concentration in reference compartment
`C. The value and ambiguity of compartment models (hydraulic analogues)
`1. One-compartment model: Vd is unique
`2. Two-compartment model: Vd is not unique23
`D. Anatomic correlates of volume of distribution
`E. Physiochemical correlates of volume of distribution24
`
`Section 5. Exponential behavior and the meaning of half-life
`A. First-order processes:
`1. Rate is proportional to concentration
`= −kC
`dC
`dt
`2. Rate is not constant, even though k is called a “rate constant”
`B. Calculation of half-life
`= 0.693
`= In 2
`k
`k
`C. k and t1/2 are independent of route of administration
`D. Logarithmic versus linear graphs (Figure 5)
`E. Interpretation and implications of half-life
`
`t1/2
`
`Time elapsed (multiples of t1/2 )
`
`Fractional completion of process
`
`1
`2
`3
`4
`
`0.5
`0.75
`0.875
`>0.90
`
`F. Implications of first-order behavior
`–t1/2, Vd, and clearance are fixed
`–After single doses, AUC (area under the curve from time = zero to “infinity”) is proportional to dose
`–At steady state, Css (steady-state concentration) is proportional to infusion rate (or dosing rate)
`
`5
`
`

`

`1185
`
`Greenblatt and Abourjaily
`
`Table 2. Continued
`
`Section 6. Clearance of drugs and mechanisms of elimination25
`A. Model-independent definition (Figure 6):
`Clearance = Dose
`AUC
`B. Units of clearance: volume/time
`Clearance cannot exceed blood flow to clearing organ
`C. Routes of clearance
`1. Hepatic clearance: chemical modification (biotransformation)
`2. Renal clearance: excretion of intact drug
`3. All other: pulmonary, biliary, fecal, intravascular (plasma enzymes)
`D. Biologic dependence of t1/2 on both Vd and clearance
`= 0.693 x Vd
`clearance
`E. The meaning of “linear” or “dose proportional” kinetics (see also section 5F)
`
`t1/2
`
`1.
`
`k
`
`t1/2
`
`Section 7. Rapid single-dose intravenous injection
`A. One-compartment model
`= −kC
`dC
`dt
`Boundary condition: at t = 0, C= C0
`C = C0 e-kt satisfies differential equation and boundary condition
`Total AUC = C0
`k
`2. If dose = D, then
`Vd = Dose
`C0
`Note: For a 1-compartment model, Vd is unique. In practice, the best time to measure Vd is at t = 0.
`3. Monoexponential decline (Figure 5)
`= 0.693
`t1/2
`= k . Vd = k . Dose
`4. Clearance = Dose
`C0
`AUC
`5. Correct biologic relation among t1/2 , Vd, and clearance:
`= 0.693 x V d
`C lear ance
`6. Graphical approach to problem solving
`B. Two-compartment model16,17(Figure 7)
`1. Simultaneous differential equations with boundary condition:
`C = C0 at t = 0
`Solution: C = Ae-αt + Be-βt
`A, B, α, β are HYBRID
`+ B
`Total AUC = A

`

`
`2. Volumes of distribution20–22
`a. “Central” compartment
`V1 = Dose
`= Dose
`A + B
`C0
`b. “Total” volume of distribution
`Vd =
`= Clearance
`Dose
`β × AUC

`
`t1 /2
`
`3. Half-lives
`a. Distribution half-life (cannot be derived from graph)
`α = 0.693

`b. Elimination half-life (can be derived from graph)
`β = 0.693
`t1 /2

`4. Clearance = dose/AUC = Vd · β
`
`6
`
`

`

`2-Compartment
`C = Ae-αt + Be-βt
`
`+ B

`
`A α
`
`Dose
`β .AUC
`= Vd . β
`
`Dose
`AUC
`In 2

`
`1186
`
`Table 2. Continued
`
`C. One-compartment approximation of the 2-compartment model
`Fundamental assumption:
`A and α (and the corresponding component of AUC) are ignored
`Consequences:
`B becomes C0
`β becomes k
`t1/2 β becomes t1/2
`
`Quantity
`
`Equation for concentration versus time
`
`Total AUC
`
`Total V d
`
`Clearance
`
`Elimination half-life dose
`
`D. Graphical approach to problem-solving
`E. Distribution versus elimination as the determinant of duration of action (Figures 3 and 7)
`
`Section 8. Multiple-dose kinetics: continuous intravenous infusion (without a loading dose) (Figure 8)
`A. Rate of attainment of steady state:
`Depends almost entirely on t1/2 (and k) as would occur with a single dose.
`Does not depend on infusion rate (abbreviation: Q).
`Inverted exponential function:
`C = Css (1 − e-kt). This is the same k as after a single dose.
`B. Determinants of the extent of accumulation (absolute steady-state concentration) (Figures 8 and 9)
`Css = Infusion rate
`Clearance
`C. Predictable consequences of changing infusion rate or stopping infusion (Figure 10)
`1. Rate of attainment of new steady state
`2. New steady-state concentration
`
`Section 9. Drug absorption and bioavailability (Figure 11)
`A. Rate of drug absorption
`1. Lag time (due to dissolution, gastric emptying, etc.)
`2. First-order absorption
`3. Factors influencing absorption rate
`4. Implications of absorption rate
`5. Slow-release preparations
`B. Completeness of absorption (fractional absorption or absolute systemic availability)
`1. Methods of assessment—intravenous data needed (Figure 12)
`F = AUC (P.O.)
`AUC (I.V.)
`AUC must be dose-normalized and extrapolated to infinity.
`2. Mechanisms of incomplete bioavailability25–29
`a. Incomplete absorption
`- Intrinsic properties of the chemical
`- Properties of the dosage form
`b. Efflux transport
`c. Presystemic extraction (first-pass metabolism)
`- Hepatic
`- Enteric (CYP3A)
`C. Bioequivalence and the pharmacopolitics of generic substitution30,31
`1. Competing political and economic influences
`2. Fundamental premise:
`Bioequivalence implies therapeutic equivalence (not the reverse)
`3. Methods of determining bioequivalence: statistical “equivalence” of Cmax, Tmax, and AUC
`
`The Journal of Clinical Pharmacology / Vol 56 No 10 2016
`
`(cid:3)
`
`1-Compartment
`C = C0e
`−kt
`(B analogous to C0,
`(cid:2)
`β analogous to k)
`C0
`(cid:2)
`k
`
`Dose
`B
`
`analogous to
`
`= Vd . k
`
`Dose
`C0
`
`Dose
`AUC
`In 2
`k
`
`(cid:3)
`
`B β
`
`analogous to
`
`7
`
`

`

`Greenblatt and Abourjaily
`
`Table 2. Continued
`
`4. Areas of uncertainty
`a. Patients versus volunteers
`b. Limits of statistical tolerance
`c. Interpretation of anecdotes
`d. Voluntary versus forced generic substitution
`e. Sustained-release preparations
`5. Special considerations for biologic therapies (biosimilars)32,33
`
`1187
`
`Section 10. Multiple-dose kinetics: discrete doses (Figure 13)
`A. There is not a unique steady-state concentration, only a mean:
`Css = AUC over dose intervel
`length of dose intervel
`B. Rate of attainment of steady-state: identical to section 8A
`C. Extent of accumulation: if each dose (D) is 100% available and given at fixed intervals (T),
`Css =
`D/T
`Clearance
`D. Interdose fluctuation (Figure 14)
`1. Estimating the degree of fluctuation
`Cmax = maximum plasma concentration over the dosage interval
`Cmin = Minimum plasma concentration over the dosage interval
`Cmax/Cmin ratio is interdose fluctuation
`2. Css stays the same as long as D/T is unchanged, but changing both D and T influences interdose fluctuation (Figure 15)
`3. Clinical benefits of “slow-release” preparations
`E. Termination of multiple dosage: the same value of k (and t1/2 ) is applicable
`F. Loading doses (DL)
`1. Determinants of when needed; benefits and disadvantages
`2. Approximate calculations:
`Given a desired Css and assuming Vd is known, DL should cause C0 for a single dose (see section 7A) to equal the target Css during continuous infusion
`(see section 8B) or Css during multiple discrete doses (see section 10C).
`3. Complications with 2-compartment model: overshoot and undershoot
`G. Relative drug accumulation: depends on the interval between doses relative to the elimination half-life
`Section 11. Nonlinear (zero-order) kinetics10,35,37 (Figure 16)
`A. Linear (first-order) kinetics (as in section 5A):
`= −kC
`dC
`dt
`Solution: C = Coe-kt
`B. Nonlinear (zero-order) kinetics
`= −k
`dC
`dt
`Solution: C = Co − kt
`C. For most affected drugs, first-order kinetic profile transitions to zero order as the concentration increases (Figure 17)
`D. Implications of transition to zero-order kinetics
`
`Section 12. Drug interactions involving altered drug clearance2,3,36–43
`A. Epidemiology of drug interactions
`B. Mechanisms of inhibition versus induction
`C. Quantitative outcome of drug interactions (Figure 18)
`D. Clinical consequences of drug interactions: depends on the quantitative magnitude of the drug interaction and the therapeutic index of the affected drug.
`Statistical significance does not imply clinical importance.
`E. In vitro prediction of clinical drug interactions: relation of inhibitor “exposure” to inhibitory “potency”
`
`Section 13. Drug therapy in vulnerable populations: the elderly44–48
`A. Epidemiology of aging
`B. Mechanisms of altered drug response in the elderly
`1. Kinetic
`2. Dynamic
`C. Consequences of impaired clearance in the elderly.
`
`Section 14. Other vulnerable or special populations
`A. Renal insufficiency
`B. Hepatic insufficiency
`C. Obesity
`D. Children
`E. Pregnancy
`
`8
`
`

`

`1188
`
`Table 2. Continued
`
`The Journal of Clinical Pharmacology / Vol 56 No 10 2016
`
`Section 15. Pharmacodynamics3,51–53
`A. Definition: time course of drug effect
`B. Approaches to measuring drug effect
`1. Fully objective (blood pressure, QT interval, serum cholesterol, etc.)
`2. Partially objective (memory tests, reaction times, pain threshold, etc.)
`3. Subjective (psychiatric endpoints)
`C. Surrogate measures of drug effect (glycated hemoglobin, T-cell count, bone mineral density, intraocular pressure, etc.)
`D. Problems with pharmacodynamic measures
`1. Effects of practice, adaptation, and time
`2. Acute and chronic tolerance
`3. Fatigue
`4. Weak connection to clinical endpoints
`E. Kinetic-dynamic modeling49–54 (Figure 19)
`1. Link between concentration and effect (linear, exponential, or sigmoid-Emax)
`2. Sources of bias
`3. Modification by effect-site entry delay
`4. Interpretation of outcome
`
`Table 3. Medical and Scientific Journals Having a Focus on Clinical
`Pharmacokinetics, Drug Metabolism, and Drug Interactions
`
`Biopharmaceutics and Drug Disposition
`British Journal of Clinical Pharmacology
`Clinical Pharmacokinetics
`Clinical Pharmacology and Therapeutics
`Clinical Pharmacology in Drug Development
`Drug Metabolism and Disposition
`European Journal of Clinical Pharmacology
`International Journal of Clinical Pharmacology
`Journal of Clinical Pharmacology
`Journal of Clinical Psychopharmacology
`Journal of Pharmaceutical Sciences
`Journal of Pharmacology and Experimental Therapeutics
`Therapeutic Drug Monitoring
`Xenobiotica
`
`Figure 4. Schematic relation between drug concentrations (green
`triangles) in circulating blood, concentrations in extracellular fluid
`(ECF) surrounding the receptor site, the extent of occupancy of
`cellular receptor sites, and subsequent pharmacologic action. If receptor
`occupancy proportionally reflects blood and ECF concentrations, then
`blood concentration may serve as a surrogate measure of drug effect.
`
`9
`
`

`

`Greenblatt and Abourjaily
`
`1189
`
`Figure 5. Plasma concentrations versus time after dosage of a drug given by rapid intravenous injection, assuming an underlying 1-compartment
`pharmacokinetic model. Each time an interval equal to the half-life elapses, the concentration falls to 50% of the value at the start of the interval.
`Left: linear concentration scale; right: logarithmic concentration scale. The same declining exponential function is applicable to both graphs (see Table
`2, section 5A–E and section 7A1–3), but the logarithmic scale transforms the exponential curve into a straight line, which can be used for graphical
`calculations. Note that a logarithmic concentration scale never goes to zero.
`
`Figure 6. Plasma concentrations of the same dose of the same drug given to different individuals. The area under the plasma concentration curve
`(AUC) is shown as the shaded region. Since clearance can be calculated as administered dose divided by AUC, the subject in the left graph has lower
`clearance (higher AUC) than the subject in the right graph (proviso: in the calculation of clearance, the “dose” represents the systemically-available
`dose, and “AUC” represents the total area under the curve from time zero to “infinity”).
`
`10
`
`

`

`1190
`
`The Journal of Clinical Pharmacology / Vol 56 No 10 2016
`
`Figure 7. Left: Plasma concentrations of a drug, consistent with a 2-compartment model, after a single intravenous dose. Right: Corresponding drug
`behavior in the 2-compartment model schematic. Point I: Immediately after the dose, the entire dose is confined to the central compartment, and the
`plasma concentration is maximal (C0 = 60). Point II: During the initial distribution phase (the “alpha” phase), plasma concentrations fall rapidly and
`extensively, due mainly to drug distribution from central to peripheral compartments. Clearance (irreversible elimination) contributes minimally to this
`initial decline. If the concentration falls below the minimal effective concentration (MEC) during the distribution phase, the distribution process may
`limit the duration of clinical action. Point III: This is the point termed distribution equilibrium. The distribution process is complete, and the ratio of central
`to peripheral compartment concentrations from this time forward will remain approximately constant. Point IV: The decline in plasma concentrations
`during the elimination phase (the “beta” phase) is relatively slow, and is due mainly to clearance (irreversible elimination). (see Table 2, section 7B)
`
`Figure 8. A continuous zero-order (fixed-rate) intravenous infusion of
`a drug obeying 1-compartment kinetics is started at time zero without
`a loading dose. The concentration rises in exponential fashion until the
`steady-state condition is reached (dashed lines). The actual steady-state
`concentration (Css) at an infusion rate of Q is 3 units. If the infusion
`rate were 2 × Q, Css would be 6 units. However the rate of attainment
`of steady state is the same in both cases, being dependent only on the
`half-life (from reference 2, with permission).
`
`Figure 9. Relation between zero-order infusion rate (Q, X axis) and
`steady-state plasma concentration (Css, Y axis) for a drug having linear
`(first-order) kinetic properties, as shown in Figure 8. Css has a direct
`linear relation to Q (see Table 2, sections 5F and 8B). The slope of the
`line is 1/clearance.
`
`11
`
`

`

`Greenblatt and Abourjaily
`
`1191
`
`Figure 10. Left: A drug is given by continuous zero-order intravenous infusion (fixed rate of Q) starting at time zero. The plasma concentration
`ascends to Css (4 units) in exponential fashion, with attainment of steady state more than 90% complete after 4 × t1/2. This is similar to Figure 8.
`Right: After steady state is reached, the infusion rate is changed (arrow), and the time scale “resets” to zero. The rate is either increased to 2 × Q,
`decreased to 0.5 × Q, or stopped altogether. The new Css correspondingly changes to 8 units, 2 units, or 0, respectively. However, another 4 or more
`multiples of t1/2 are needed for the new steady state to be reached.
`
`Figure 11. Plasma concentrations of a drug after oral dosage at
`time zero. After a lag time (Tlag), plasma concentrations begin to rise.
`During this “absorption” phase, rates of drug entry into blood due to
`absorption exceed rates of elimination due to clearance. The maximum
`concentration (Cmax = 7 units) is reached at time Tmax (1 hour after
`dosage); at this point, instantaneous absorption and elimination rates are
`equal. Concentrations then fall, indicating that elimination rates exceed
`absorption rates. The area under the plasma concentration curve (AUC)
`is used as a surrogate for the extent of absorption (from reference 2,with
`permission).
`
`Figure 12. Mean plasma concentrations of midazolam after single
`intravenous and oral doses administered to healthy volunteers29 (*con-
`centrations were normalized to a 2-mg dose). Based on the relationship
`in section 9B1, in Table 2, the absolute bioavailability of oral midazolam
`(F) was calculated as 0.29,indicating that only 29% of an oral dose actually
`reaches the systemic circulation (from reference 2, with permission).
`
`12
`
`

`

`1192
`
`The Journal of Clinical Pharmacology / Vol 56 No 10 2016
`
`Figure 13. A drug is administered as discrete oral doses at intervals equal to the half-life. Six consecutive doses were given, after which the drug
`was discontinued. The dashed line is the hypothetical curve if the same dosing rate were administered by continuous zero-order intravenous infusion.
`Note that the rate of attainment of steady state is the same in both cases.
`
`Figure 14. A dosage interval at steady state. The elimination half-life
`is assumed to be 24 hours, and the dosage interval is also 24 hours
`(as in Figure 13). The plasma concentration starts at Cmin, increases to
`Cmax, then falls again to Cmin. The mean steady-state concentration (Css,
`dashed line) is the area under the curve for 1 dose interval divided by
`the length of the interval. The interdose fluctuation is calculated as the
`Cmax/Cmin ratio. (see Table 2, section 10A–D).
`
`Figure 15. Serum concentrations of a drug at steady state if the drug
`is given using 3 different dosing schedules: either 500 mg every 24 hours,
`250 mg every 12 hours, or 125 mg every 6 hours. In each case the mean
`steady-state concentration (Css) is the same, since the [(dose)/(dose
`interval)] ratio does not change. However, the interdose fluctuation
`becomes smaller as the dose interval becomes smaller. With the once-
`daily dosing schedule, the interdose fluctuation is large, and the plasma
`concentration falls outside the therapeutic range just after and just
`before each

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket